Field of the Invention
[0001] The present invention relates to a method of inhibiting proliferation of prostate
cancer cells, such as in a method of treating prostate cancer. In particular, the
invention relates to a method of inhibiting proliferation of androgen-independent
prostate cancer cells.
Background of the invention
[0002] Prostate cancer occurs frequently in men, is currently the second most common cause
of cancer-related death and incidence is growing, Prostatectomy is useful in the treatment
of patients with cancer confined to the prostate. Androgen ablation therapy is used
in cases where cancer cells still require androgens for growth but have spread beyond
the prostate. To date, however, there is no effective treatment for metastatic Androgen-Independent
Prostate Cancer (AIPC).
[0003] Our understanding of the aetiology of prostate cancer is limited and unlike certain
other cancers, little progress has been made in elucidating its cause. Efforts have
been made to identify genes responsible for familial prostate cancer. At least seven
chromosomal loci have been reported, however the genes responsible for prostate cancer
in all these loci have not yet been identified. Although an inherited genetic predisposition
occurs in only 5-10% of cases, it is possible that identification of germline mutations
may shed light on sporadic cases as both forms share the same histopathological features.
The majority of researchers have focused on somatic defects in sporadic prostate cancer.
Classical cytogenetic studies are difficult to apply to solid tumours and so far no
consistent chromosomal changes have been observed. Although comparative genome hybridisation
and loss of heterozygosity analysis have shown both gain and loss of genomic DNA,
the majority of genes involved are still unknown. Oncogenes and tumour suppressor
genes known to be associated with other malignancies have a remarkably low frequency
of mutation or deletion in prostate cancer. Using technologies that compare the steady-state
mRNA levels between normal and cancerous prostate, a list of genes have been revealed
to be either over or underexpressed in prostate cancer tissue or cell-lines. Although
proteomics and tissue array approaches are now being used, relatively few genes have
yet been verified to be differentially expressed in a reasonable number of specimens
at the protein level. Direct evidence for the importance of these differentially expressed
genes in prostate cancer initiation or progression is lacking. As a result, although
progress is rapid, the application of this new knowledge in controlling mortality
and morbidity from prostate cancer is slow at present.
[0004] Emerging evidence from epidemiological studies indicates a strong association between
prostate cancer risk and total fat intake (
Kolonel et al., 1999 J. Natl Cancer Inst. 91: 414), although the biochemical link between dietary lipids and genesis of prostate cancer
remains unclear. Previous studies have demonstrated that both cyclooxygenase (COX)
and lipoxygenase (LOX) products of arachidonic acid metabolism, the prostaglandins
(PG), and hydroxyeicosatetraenoic acids (HETEs) respectively, contribute to formation
and/or progression of prostate cancer. They are implicated in promotion of tumour
cell proliferation, motility, invasion and metastasis, and induction of angiogenesis
both
in vitro and in animal models. Interestingly, arachidonic acid levels are lower in malignant
than benign (BPH) prostate tissue while PG and HETE synthesis from labelled arachidonic
acid is significantly increased. However, the activity of arachidonic acid mobilising
enzymes phospholipase A
2 (PLA
2) and fatty acyl-CoA lysophosphatidylcholine acyltransferase, are also increased,
suggesting an increased flux of arachidonic acid through the COX and LOX pathways.
[0005] PLA
2 constitutes a large and diverse family of enzymes that catalyse the hydrolysis of
membrane phospholipids at the
sn-2 position to release fatty acids and lysophospholipids. PLA
2 enzymes are classified according to their source and their cellular location (i.e
secreted PLA
2 enzymes (sPLA
2s) or cytosolic PLA
2 enzymes (cPLA
2s)). A review of the classification and characterisation of the expanding superfamily
of PLA
2 enzymes had been published by
Six and Dennis (2000) Biochim. Biophys. Acta 1488:1-19.
[0006] sPLA
2-IIA is elevated in prostate cancer (
Graff et al., 2001, Clin. Cancer Res. 7: 3857-3861;
Jiang et al., 2002, Am. J. Pathol. 160: 667-671) and enhanced sPLA
2-IIA expression has been inversely related to 5-year patient survival (Graff et al.,
2001). In addition, the chromosomal location of several sPLA
2 genes including sPLA
2-IIA (1p35-ter), overlaps with one prostate cancer susceptibility locus CAPB (
Nwosu et al., 2001, Human Mol. Genet. 10: 2313-2318). To date, however, there has been no evidence to show that sPLA
2-IIA is involved in prostate tumorigenesis.
Summary of the invention
[0007] We have now shown by immunohistochemistry in patients following androgen ablation
therapy, that Type IIA secreted phospholipase A
2 (sPLA
2-IIA) is elevated in androgen-independent tumour cells relative to benign glands,
while cytosolic phospholipase A
2-α (cPLA
2-α) levels are unchanged.
[0008] We have also found that treatment of prostate cancer cells with sPLA
2-IIA potently increases proliferation of the cells, and that this proliferative effect
is blocked by the addition of selective inhibitors of both sPLA
2-IIA and cPLA
2-α enzymes.
[0009] This provides evidence for the first time of the direct role of PLA
2 in the proliferation of prostate cancer cells and identifies this class of enzymes
as an important therapeutic target for the treatment of prostate cancer.
[0010] Accordingly, in a first aspect the present invention provides a method of inhibiting
or reducing the proliferation of prostate cancer cells, the method comprising administering
to the cells a PLA
2 inhibitor.
[0011] In a second aspect the present invention provides a method for the treatment of prostate
cancer, the method comprising administering to a subject in need thereof a PLA
2 inhibitor.
[0012] In a preferred embodiment of the first and second aspects, the prostate cancer cells
are androgen independent prostate cancer (AIPC) cells.
[0013] In accordance with the first and second aspects of the present invention, the PLA
2 inhibitor may inhibit any PLA
2 enzyme. Preferably, the inhibitor inhibits an enzyme selected from group 1B, IIA,
IID, IIE, IIF, III, IV, V, and X PLA
2 enzymes.
[0014] In one embodiment, the inhibitor is a cPLA
2-α inhibitor. For example, the inhibitor may be pyrrolidine-1, a substituted pyrimidine
as described in
WO 00/27824, a 9,10-dihydro-9,10-ethanoanthracene derivative as described in
WO 99/15493, an azalomycin inhibitor as described in
JP12119292, an arylsulfonamide as described in
WO 98/25893, an indole-2-carboxylic acid derivative as described in
WO 98/05637, an indole derivative as described in
WO 98/08818,
WO 99/43651,
WO 99/43654 or
WO 99143672, a 3-sulfanyl-propane- 1,2-diol derivative as described in
JP12038380, a heterocyclic compound such as those described in
WO 00/34254 or an oxazolidinedione or thiazolidinedione derivative as described in
WO 97/05135 or
WO 98/33797.
[0015] In another embodiment, the inhibitor is an sPLA
2-IIA inhibitor. For example, the inhibitor may be a benzoic acid as described in
JP8325154, a 6-aza-spiro-[4,5]-decane derivative as described in
JP9110835, amide derivatives of fatty acids such as those described in
WO 00/00220 and
WO 95/19959, (arylsulfonamidophnoxy) benzoic acids such as those described in
WO 97/35567, fatty acid derivatives such as those described in
WO 97/38966, tetronic acids such as those described in
JP0045740, dinitrogen heterocyclic compounds such as those described in
WO 98/05332, oxadiazine and thioxadiazine derivatives such as those describe in
WO 00/71118, sulfonylaminopyrazoles such as those described in
WO 98/24437, (indol-3-yl)acetamide inhibitors described in
EP 839806,
EP 950657,
EP 952149 and
WO 00/07590. (indol-3-yl) oxoacetamides such as those described in
EP 675110,
WO 98/37069,
WO 99/59999,
WO 99/51605,
WO 00/07591,
WO 00/37358 and
WO 00/00201, or derivatives such as those described in
WO 99/21545,
WO 99/21546 and
WO 99/21559.
[0016] In a particularly preferred embodiment the inhibitor is a conformationally constrained
molecule derived from a peptide consisting essentially of amino acid residues 70 to
74 of a human sPLA
2-IIA protein, or the equivalent residues in other sPLA
2-IIA proteins.
[0017] Preferably, the conformationally constrained molecule is a peptide, more preferably
a cyclic peptide.
[0018] In a preferred embodiment, the conformationally constrained peptide is a cylic peptide
of the hollowing formula:
A1-A2-A3-A4-A5
in which
A1 is F or Y or W or 2Nap
A2 is L or I
A3 is S or T
A4 is F or Y or W or 2Nap
A5 is R or K.
[0019] In a further preferred embodiment of the present invention, the peptide is selected
from the group consisting of cFLSYK, cFLSYR and c(2NapA)LS(2NapA)R.
[0020] When used herein the term "cFLSYK" means "cyclic FLSYK", "cFLSYR" means "cyclic FLSYR"
and "c(2NapA)LS(2NapA)R" means "cyclic (2Nap)LS (2Nap)R". The term "2NapA" is an abbreviation
for 2-naphthylalanine.
[0021] In a further preferred embodiment, the peptide comprises D-amino acids and has a
sequence which corresponds to the reverse sequence of a peptide according to the first
aspect of the present invention.
[0022] In a preferred embodiment of the present invention, the method involves administration
of a sPLA
2-IIA inhibitor in conjunction with a cPLA
2-
α inhibitor.
[0023] In yet a further aspect the present invention provides the use of a PLA
2 inhibitor in the manufacture of a medicament for the treatment of prostate cancer.
[0024] In yet a further aspect the present invention provides the use of a conformationally
constrained molecule derived from a peptide consisting essentially of amino acid residues
70 to 74 of a human sPLA
2-IIA protein, or the equivalent residues in other sPLA
2-IIA proteins, in the manufacture of a medicament for the treatment of prostate cancer.
[0025] In yet another aspect the present invention provides a method for detecting prostate
cancer or a metastasis thereof in a subject, said method comprising:
determining the level of PLA2 mRNA expressed in a test sample from said subject; and
comparing the level of PLA2 mRNA determined at (i) to the level of PLA2.mRNA expressed in a comparable sample from a healthy or normal individual,
wherein a level of PLA2 mRNA at (i) that is enhanced in the test sample relative to the comparable sample
from the normal or healthy individual is indicative of the presence of a cancer cell
in said subject.
[0026] In yet another aspect the present invention provides a method for detecting prostate
cancer or a metastases thereof in a subject, said method comprising:
determining the level of a PLA2 polypeptide in a test sample from said subject; and
comparing the level of PLA2 polypeptide determined at (i) to the level of said PLA2 polypeptide in a comparable sample from a healthy or normal individual,
wherein a level of said PLA2 polypeptide at (i) that is enhanced in the test sample relative to the comparable
sample from the normal or healthy individual is indicative of the presence of a cancer
cell in said subject.
[0027] In yet another aspect the present invention provides a method of assessing the predisposition
of a subject to prostate cancer, the method comprising the step of determining the
presence of a polymorphism or an epigenetic change in a PLA
2 gene of the subject.
[0028] In the methods for detecting prostate cancer and the methods of assessing the predisposition
of a subject to prostate cancer disclosed herein, the prostate cancer cells may be
androgen independent prostate cancer (AIPC) cells. In any of these methods, the PLA
2 may be cPLA
2-α or sPLA
2-IIA.
Brief description of the Figures
[0029]
Figure 1. Effect of androgen ablation therapy on sPLA2-IIA protein expression. Prostate tissue from patients following radical prostatectomy
either with (N = 25) or without (N=50) prior androgen ablation therapy were examined
for sPLA2-IIA protein expression by immunohistochemistry as described in Materials and Methods.
A. Adjacent sections from a patient without androgen-ablation therapy showing normal
glands. B. Adjacent sections from a patient following androgen ablation therapy showing
both normal and neoplastic tissue. Left panel stained with haematoxilin and eosin,
right panel stained with anti-sPLA2-IIA antibody. Magnification, x 40. C. sPLA2-IIA expression in normal (open boxes) or neoplastic (closed boxes) tissue from patients
with (+) or without (-) androgen ablation therapy was graded on a 3-point score, (1;
0 - 33% tissue positive, 2; 33 - 66% tissue positive and 3; >66% tissue positive).
*; P < 0.05 vs no androgen ablation therapy treated benign glands by Chi-Square analysis.
Figure 2. Effect of exogenous addition of sPLA2-IIA on prostate cell growth. LNCaP cells were grown in RPMI with 5% FCS and treated
for 72 hours in media containing (A) increasing concentrations of sPLA2-IIA alone (closed bars) or increasing concentrations of the activity-dead sPLA2-IIA mutant H48Q. (B) a fixed concentration of sPLA2-IIA and increasing concentrations of the sPLA2-IIA inhibitor cFLSYR. Cell number relative to untreated control cells was determined
by the MTS assay as described in Materials and Methods. Data are Mean ± SD of quadruplicate
determinations normalised to 100% for untreated cells. OD495 of unstimulated cells was 0.29 ±0.06 for sPLA2-IIA and 0.24 ± 0.03 for the sPLA2-IIA mutant experiments in panel A and 0.24 ± 0.03 for panel B. * p<0.05 vs untreated
control by one-way ANOVA. Data are representative of three separate experiments. (C).
LNCaP cells were grown as above and stimulated for 72 hours with sPLA2-IIA (1 nM) in the absence (dotted lines) and presence (solid lines) of the sPLA2-IIA inhibitor cFLSYR (100 nM). Cells (1x106) were assayed by flow cytometry following treatment with propidium iodide as described
in Materials and Methods. Statistical analysis was performed on 10,000 events per
sample. Data are representative of three separate experiments.
Figure 3. Effect of inhibition of cPLA2-α on sPLA2-IIA-dependent proliferation. LNCaP cells were grown in RPMI containing 5% FCS and
stimulated for 72 hours with sPLA2-IIA (1 nM) in the presence (hatched bars) and absence (closed bar) of increasing
concentrations of the cPLA2-α-selective inhibitor pyrrolidine-1. Cell number was determined by the MTS assay
as described in Materials and Methods. Data are mean ± SD of quadruplicate determinations
expressed as percentages relative to untreated control cells (100%) (open bar). OD495 of untreated cells was 0.33 ± 0.03. In the presence of pyrrolidine-1 (5 uM) alone,
OD495 was 0.38 ± 0.04 * p<0.05 vs untreated control by one-way ANOVA.
Figure 4. Effect of sPLA2-IIA inhibition on unstimulated prostate cancer cells. (A) The endogenous expression
of sPLA2-IIA mRNA was evaluated by RT-PCR in three unstimulated prostate cancer cell lines
(LNCaP, DU145 and PC-3) grown in RPMI containing 5% FCS. HPRT was used to as a positive
control for RNA integrity and loading. (B) Cells were grown in RPMI with 5% FCS were
then treated for 72 hours in the presence and absence of the sPLA2-IIA inhibitor c(2Nap)LS(2Nap)R and cell number determined by MTS assay as described
in Materials and Methods. Data are mean ± SD of quadruplicate determinations expressed
as percentages relative to untreated control cells (100%). LNCaP, closed bars, OD495 untreated cells 0.50 ± 0.03; DU145, open bars, OD495 untreated cells 0.73 ± 0.05; PC-3, hatched bars, OD495 untreated cells, 0.66 ±0.01. * p<0.05 vs untreated control by one-way ANOVA.
Detailed description of the invention
[0030] Unless defined otherwise, all technical and scientific terms used herein have the
same meaning as commonly understood by one of ordinary skill in the art (e.g. in cell
culture, molecular genetics, nucleic acid chemistry, hybridization techniques and
biochemistry). Standard techniques are used for molecular, genetic and biochemical
methods (see generally,
Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd ed. (1989) Cold Spring
Harbor Laboratory Press, Cold Spring Harbor, N.Y. and
Ausubel et al., Short Protocols in Molecular Biology (1999) 4th Ed, John Wiley & Sons,
Inc. - and the full version entitled Current Protocols in Molecular Biology, which are
incorporated herein by reference) and chemical methods.
[0031] Throughout this specification the word "comprise", or variations such as "comprises"
or "comprising", will be understood to imply the inclusion of a stated element, integer
or step, or group of elements, integers or steps, but not the exclusion of any other
element, integer or step, or group of elements, integers or steps.
PLA2 Enzymes
[0032] The methods of the present invention encompass targeting any PLA
2 enzyme. A review of the classification and characterisation of the expanding superfamily
of PLA
2 enzymes had been published by
Six and Tennis (2000) Biochim. Biophys. Acta 1488:1-19, and the present methods contemplate targeting all of these enzymes.
[0033] In preferred embodiments of the present invention, the PLA
2 enzyme is sPLA
2-IIA or cPLA
2-α. Preferably, the PLA
2 enzyme is a human enzyme.
[0034] The nucleic acid and protein sequences for human sPLA
2-IIA are shown in SEQ ID NOs:1 and 3 respectively. The nucleic acid and protein sequences
for human cPLA
2-α are shown in SEQ ID NOs:2 and 4 respectively.
PLA2 Inhibitors
Protein or Peptide inhibitors
[0035] In one embodiment, peptidyl PLA
2 inhibitors are chemically or recombinantly synthesized as oligopeptides (approximately
10-25 amino acids in length) derived from the a PLA
2 sequence (for example, SEQ ID NO:3 or 4). Alternatively, PLA
2 fragments are produced by digestion of native or recombinantly produced PLA
2 by, for example, using a protease, e.g., trypsin, thermolysin, chymotrypsin, or pepsin.
Computer analysis (using commercially available software, e.g. MacVector, Omega, PCGene,
Molecular Simulation, Inc.) is used to identify proteolytic cleavage sites. The proteolytic
or synthetic fragments can comprise as many amino acid residues as are necessary to
partially or completely inhibit PLA
2 function. Preferred fragments will comprise at least 5, 10, 15, 20, 25, 30, 35, 40,
45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more amino acids in length.
[0036] Protein or peptide inhibitors may also be dominant-negative mutants of PLA
2. The term "dominant-negative mutant" refers to a PLA
2 polypeptide that has been mutated from its natural state and that interacts with
a protein that PLA
2 normally interacts with thereby preventing endogenous native PLA
2 from forming the interaction.
Anti-PLA2 Antibodies
[0037] The term "antibody" as used in this invention includes intact molecules as well as
fragments thereof, such as Fab, F(ab')2, and Fv which are capable of binding an epitopic
determinant of PLA
2. These antibody fragments retain some ability to selectively bind with its antigen
and are defined as follows:
(1) Fab, the fragment which contains a monovalent antigen-binding fragment of an antibody
molecule can be produced by digestion of whole antibody with the enzyme papain to
yield an intact light chain and a portion of one heavy chain;
(2) Fab', the fragment of an antibody molecule can be obtained by treating whole antibody
with pepsin, followed by reduction, to yield an intact light chain and a portion of
the heavy chain; two Fab' fragments are obtained per antibody molecule;
(3) (Fab')2, the fragment of the antibody that can be obtained by treating whole antibody
with the enzyme pepsin without subsequent reduction; F(ab)2 is a dimer of two Fab'
fragments held together by two disulfide bonds;
(4) Fv, defined as a genetically engineered fragment containing the variable region
of the light chain and the variable region of the heavy chain expressed as two chains;
and
(5) Single chain antibody ("SCA"), defined as a genetically engineered molecule containing
the variable region of the light chain, the variable region of the heavy chain, linked
by a suitable polypeptide linker as a genetically fused single chain molecule.
[0039] Antibodies of the present invention can be prepared using intact PLA
2 or fragments thereof as the immunizing antigen. A peptide used to immunize an animal
can be derived from translated cDNA or chemical synthesis and is purified and conjugated
to a carrier protein, if desired. Such commonly used carriers which are chemically
coupled to the peptide include keyhole limpet hemocyanin (KLH), thyroglobulin, bovine
serum albumin (BSA), and tetanus toxoid. The coupled peptide may then be used to immunize
the animal (e.g., a mouse or a rabbit).
[0040] If desired, polyclonal antibodies can be further purified, for example, by binding
to and elution from a matrix to which the peptide to which the antibodies were raised
is bound. Those of skill in the art will know of various techniques common in the
immunology arts for purification and/or concentration of polyclonal antibodies, as
well as monoclonal antibodies (See for example,
Coligan, et al., Unit 9, Current Protocols in immunology, Wiley Interscience, 1991, incorporated by reference).
[0041] Monoclonal antibodies may be prepared using any technique which provides for the
production of antibody molecules by continuous cell lines in culture, such as, for
example, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma
technique (
Kohler et al. Nature 256, 495-497, 1975;
Kozbor et al., J. lmmunol. Methods 81, 31-42, 1985;
Cote et al., Proc. Natal. Acad. Sci. USA 80, 2026-2030, 1983;
Cole et al., Mol. Cell Biol. 62, 109-120, 1984).
[0042] Methods known in the art allow antibodies exhibiting binding for PLA
2 to be identified and isolated from antibody expression libraries. For example, a
method for the identification and isolation of an antibody binding domain which exhibits
binding to PLA
2 is the bacterio-phage a vector system. This vector system has been used to express
a combinatorial library of Fab fragments from the mouse antibody repertoire in Escherichia
coli (
Huse, et al., Science, 246:1275-1281, 1989) and from the human antibody repertoire (
Mullinax, et al., Proc. Nat. Acad. Sci., 87:8095-8099, 1990). This methodology can also be applied to hybridoma cell lines expressing monoclonal
antibodies with binding for a preselected ligand. Hybridomas which secrete a desired
monoclonal antibody can be produced in various ways using techniques well understood
by those having ordinary skill in the art and will not be repeated here. Details of
these techniques are described in such references as
Monoclonal Antibodies-Hybridomas: A New Dimension in Biological Analysis, Edited
by Roger H. Kennett, et al., Plenum Press, 1980; and
U.S. 4,172,124, incorporated by reference.
Antisense compounds
[0044] The term "antisense compounds" encompasses DNA or RNA molecules that are complementary
to at least a portion of a PLA
2 mRNA molecule (
Izant and Weintraub, Cell 36:1007-15, 1984;
Izant and Weintraub, Science 229(4711):345-52, 1985) and capable of interfering with a post-transcriptional event such as mRNA translation.
Antisense oligomers complementary to at least about 15 contiguous nucleotide of PLA
2-encoding mRNA are preferred, since they are easily synthesized and are less likely
to cause problems than larger molecules when introduced into the target PLA
2-producing cell. The use of antisense methods is well known in the art (
Marcus-Sakura, Anal. Biochem. 172: 289, 1988). Preferred antisense nucleic acid will comprise a nucleotide sequence that is complementary
to at least 15 contiguous nucleotides of a sequence encoding the amino acid sequence
set forth in SEQ ID NO:3 or 4.
Catalytic nucleic acids
[0045] The term catalytic nucleic acid refers to a DNA molecule or DNA-containing molecule
(also known in the art as a "DNAzyme") or an RNA or RNA-containing molecule (also
know as a "ribozyme") which specifically recognizes a distinct substrate and catalyzes
the chemical modification of this substrate. The nucleic acid bases in the catalytic
nucleic acid can be bases A, C, G, T and U, as well as derivatives thereof. Derivatives
of these bases are well known in the art.
[0046] Typically, the catalytic nucleic acid contains an antisense sequence for specific
recognition of a target nucleic acid, and a nucleic acid cleaving enzymatic activity
(also referred to herein as the "catalytic domain"). To achieve specificity, preferred
ribozymes and DNAzymes will comprise a nucleotide sequence that is complementary to
at least about 12-15 contiguous nucleotides of a sequence encoding the amino acid
sequence set forth in SEQ ID NO:3 or 4.
[0047] The types of ribozymes that are particularly useful in this invention are the hammerhead
ribozyme (Haseloff and Gerlach 1988, Perriman et al., 1992) and the hairpin ribozyme
(Shippy et al., 1999).
[0048] The ribozymes of this invention and DNA encoding the ribozymes can be chemically
synthesized using methods well known in the art. The ribozymes can also be prepared
from a DNA molecule (that upon transcription, yields an RNA molecule) operably linked
to an RNA polymerase promoter, e.g., the promoter for T7 RNA polymerase or SP6 RNA
polymerase. Accordingly, also provided by this invention is a nucleic acid molecule,
i.e., DNA or cDNA, coding for the ribozymes of this invention. When the vector also
contains an RNA polymerase promoter operably linked to the DNA molecule, the ribozyme
can be produced in vitro upon incubation with RNA polymerase and nucleotides. In a
separate embodiment, the DNA can be inserted into an expression cassette or transcription
cassette. After synthesis, the RNA molecule can be modified by ligation to a DNA molecule
having the ability to stabilize the ribozyme and make it resistant to RNase. Alternatively,
the ribozyme can be modified to the phosphothio analog for use in liposome delivery
systems. This modification also renders the ribozyme resistant to endonuclease activity.
RNA inhibitors
[0049] dsRNA is particularly useful for specifically inhibiting the production of a particular
protein. Although not wishing to be limited by theory, Dougherty and Parks (1995)
have provided a model for the mechanism by which dsRNA can be used to reduce protein
production. This model has recently been modified and expanded by Waterhouse et al.
(1998). This technology relies on the presence of dsRNA molecules that contain a sequence
that is essentially identical to the mRNA of the gene of interest, in this case an
mRNA encoding a PLA
2 protein. Conveniently, the dsRNA can be produced in a single open reading frame in
a recombinant vector or host cell, where the sense and anti-sense sequences are flanked
by an unrelated sequence which enables the sense and anti-sense sequences to hybridize
to form the dsRNA molecule with the unrelated sequence forming a loop structure. The
design and production of suitable dsRNA molecules targeted agasint PLA
2 is well within the capacity of a person skilled in the art, particularly considering
Dougherty and Parks (1995), Waterhouse et al. (1998),
WO 99/32619,
WO 99/53050,
WO 99/49029, and
WO 01/34815.
[0050] As used herein, the terms "small interfering RNA", and "RNAi" refer to homologous
double stranded RNA (dsRNA) that specifically targets a gene product, thereby resulting
in a null or hypomorphic phenotype. Specifically, the dsRNA comprises two short nucleotide
sequences derived from the target RNA encoding PLA
2 and having self-complementarity such that they can anneal, and interfere with expression
of a target gene, presumably at the post-transcriptional level. RNAi molecules are
described by
Fire et al., Nature 391, 806-811, 1998, and reviewed by
Sharp, Genes & Development, 13, 139-141, 1999).
Small molecule inhibitors
Peptides and peptide analogues
[0052] In preferred embodiments, the methods of the present invention involve the administration
of conformationally constrained molecules derived from a peptide consisting essentially
of amino acid residues 70 to 74 of a human sPLA
2-IIA protein, or the equivalent residues in other sPLA
2-IIA proteins.
[0053] In general, reference to amino acid residues 70 to 74 of the human sPLA
2-IIA protein is taken to include reference to the equivalent residues in other sPLA
2-IIA proteins, such as orthologues of human sPLA
2-IIA.
[0054] The term "conformationally constrained molecules" means conformationally constrained
peptides and conformationally constrained peptide analogues and derivatives.
[0055] Thus the conformationally constrained molecules according to the present invention
include conformationally constrained peptides consisting essentially of residues 70
to 74 of the human sPLA
2-IIA protein, and analogues and derivatives thereof.
[0056] The term "analogues" refers to molecules having a chemically analogous structure
to the naturally occurring alpha-amino acids present as residues 70 to 74 of the human
sPLA
2-IIA protein. Examples include molecules containing
gem-diaminoalkyl groups or alklylmalonyl groups.
[0057] The term "derivatives" includes alpha amino acids wherein one or more side groups
found in the naturally occurring alpha-amino acids present as residues 70 to 74 of
human sPLA
2-IIA protein have been modified. Thus, for example the naturally-occurring amino acids
present in residues 70 to 74 of the human SPLA
2-IIA protein may be replaced with a variety of uncoded or modified amino acids such
as the corresponding D-amino acid or N-methyl amino acid. Other modifications include
substitution of hydroxyl, thiol, amino and carboxyl functional groups with chemically
similar groups.
[0058] The present invention encompasses the use of conformationally constrained peptidomimetics
of the biologically active human sPLA
2-IIA peptide (amino acid residues 70 to), i.e. analogues and derivatives which mimic
the activity of said peptide and are therefore capable of inhibiting the sPLA
2-IIA dependent proliferation of prostate cancer cells. These peptidomimetics are preferably
substantially similar in both three-dimensional shape and biological activity to the
specific sPLA
2-IIA peptides described herein. Substantial similarity means that the geometric relationship
of groups in the peptide that react with the sPLA
2-IIA enzyme is preserved and at the same time, that the peptidomimetic will inhibit
the sPLA
2-IIA dependent proliferation of prostate cancer cells.
[0059] A peptidomimetic is a molecule that mimics the biological activity of a peptide but
is no longer peptidic in chemical nature. By strict definition, a peptidomimetic is
a molecule that no longer contains any peptide bonds (that is, amide bonds between
amino acids). However, the term peptide mimetic is sometimes used to describe molecules
that are no longer completely peptidic in nature, such as pseudo-peptides, semi-peptides
and peptoids. Whether completely or partially non-peptide, peptidomimetics for use
in the methods of the invention provide a spatial arrangement of reactive chemical
moieties that closely resembles the three-dimensional arrangement of active groups
in the peptide on which the peptidomimetic is based. As a result of this similar active-site
geometry, the peptidomimetic has effects on biological systems which are similar to
the biological activity of the peptide.
[0060] There are clear advantages for using a mimetic of a given peptide rather than the
peptide itself, because peptides commonly exhibit two undesirable properties: (1)
poor bioavailability; and (2) short duration of action. Peptide mimetics offer an
obvious route around these two major obstacles, since the molecules concerned are
small enough to be both orally active and have a long duration of action. There are
also considerable cost savings and improved patient compliance associated with peptide
mimetics, since they can be administered orally compared with parenteral administration
for peptides. Furthermore, peptide mimetics are much cheaper to produce than peptides.
[0061] Suitable peptidomimetics based on residues 70 to 74 of human sPLA
2-IIA peptides and having similar biological activities, and therefore similar therapeutic
utilities, can be developed using readily available techniques. Thus, for example,
peptide bonds can be replaced by non-peptide bonds that allow the peptidomimetic to
adopt a similar structure, and therefore biological activity, to the original peptide.
Further modifications can also be made by replacing chemical groups of the amino acids
with other chemical groups of similar structure. The development of peptidomimetics
derived from sPLA
2-IIA peptides based on residues 70 to 74 of human sPLA
2-IIA can be aided by determining the tertiary structure of the original peptide by
NMR spectroscopy, crystallography and/or computer-aided Molecular modelling. These
techniques aid in the development of analogues/derivatives of higher potency and/or
greater bioavailability and/or greater stability than the original peptide (
Dean, 1994, BioEssays, 16: 683-687;
Cohen and Shatzmiller, 1993, J. Mol. Graph., 11:166-173;
Wiley and Rich, 1993, Med. Res. Rev., 13: 327-384;
Moore, 1994, Trends Pharmacol. Sci., 15: 124-129;
Hruby, 1993, Biopolymers, 33: 1073-1082;
Bugg et al., 1993, Sci. Am., 269: 92-98.
[0062] Information on the structure of an sPLA
2-IIA peptide consisting essentially of residues 70 to 74 of human sPLA
2-IIA can be used to search three-dimensional databases to identify molecules having
a similar structure, using programs such as MACCS-3D and ISIS/3D (molecular Design
Ltd., San Leandro, CA), ChemDBS-3D (Chemical Design Ltd., Oxford, U.K.), and Sybyl/3DB
Unity (Tripos Associates, St. Louis, MO).
[0063] Databases of chemical structures are available from a number of sources including
Cambridge Crystallographic Data Centre (Cambridge, U.K.), Chemical Abstracts Service
(Columbus, OH), and ACD-3D (Molecular Design Ltd).
[0064] De novo design programs include Ludi (Accelrys), Leapfrog (Tripos Associates) and Aladdin
(Daylight Chemical Information Systems, Irvine, CA).
[0065] Those skilled in the art will recognize that the design of a mimetic may require
sight structural alteration or adjustment of a chemical structure designed or identified
using these databases.
[0066] Peptide derivatives and peptidomimetic compounds based on amino acid residues 70
to 74 of human sPLA
2-IIA can be tested to determine whether they are capable of inhibiting sPLA
2-IIA dependent proliferation of prostate cancer cells using the assay described herein.
Preferred peptide derivatives and peptidomimetics have at least 90%, preferably at
least the same antiproliferative activity toward prostate cancer cells as cFLSYR.
It is also preferred that peptide derivatives and peptidomimetics specifically inhibit
sPLA
2-IIA.
[0067] The molecules, such as peptides, used in the methods of the present invention are
conformationally constrained. Conformational constraint refers to the stability and
preferred conformation of the three-dimensional shape assumed by a peptide. Conformational
constraints include local constraints, involving restricting the conformational mobility
of a single residue in a peptide; regional constraints, involving restricting the
conformational mobility of a group of residues, which residues may form some secondary
structural unit; and global constraints, involving the entire peptide structure.
[0068] The active conformation of a peptide may be stabilized by a covalent modification,
such as cyclization or by incorporation of gamma-lactam or other types of bridges.
For example, side chains can be cyclized to the backbone so as create a L-gamma-lactam
moiety on each side of the interaction site. See, generally,
Hruby et al., 1992, "Applications of Synthetic Peptides," in Synthetic Peptides: A
User's Guide: 259-345 (W. H. Freeman & Co.). Cyclization also can be achieved, for example, by formation of cystine bridges,
coupling of amino and carboxy terminal groups of respective terminal amino acids,
or coupling of the amino group of a Lys residue or a related homolog with a carboxy
group of Asp, Glu or a related homolog. Coupling of the alpha-amino group of a polypeptide
with the epsilon-amino group of a lysine residue, using iodoacetic anhydride, can
be also undertaken. See, for example,
Wood and Wetzel, 1992, Int'I J. Peptide Protein Res. 39: 533-39.
[0069] Another approach described in
US 5,891,418 is to include a metal-ion complexing backbone in the peptide structure. Typically,
the preferred metal-peptide backbone is based on the requisite number of particular
coordinating groups required by the coordination sphere of a given complexing metal
ion. In general, most of the metal ions that may prove useful have a coordination
number of four to six. The nature of the coordinating groups in the peptide chain
includes nitrogen atoms with amine, amide, imidazole, or guanidino functionalities;
sulfur atoms of thiols or disulfides; and oxygen atoms of hydroxy, phenolic, carbonyl,
or carboxyl functionalities. In addition, the peptide chain or individual amino acids
can be chemically altered to include a coordinating group, such as for example oxime,
hydrazino, sulthydryl, phosphate, cyano, pyridino, piperidino, or morpholino.
[0070] A further approach approach is to use bifunctional crosslinkers, such as N-succinimidyl
3-(2 pyridyldithio) propionate, succinimidyl 6-[3-(2 pyridyldithio) propionamido]
hexanoate, and sulfosuccinimidyl 6-[3-(2 pyridyldithio) propionamido]hexanoate (see
US Patent 5,580,853).
[0071] Techniques for chemically synthesising the peptides and derivatives described above
are described in the above references and also reviewed by
Borgia and Fields, 2000, TibTech 18: 243-251 and described in detail in the references contained therein.
Diagnostic methods
[0072] The present invention also encompasses nucleic acid-based methods and protein-based
methods for diagnosing prostate cancer in humans and other mammals.
[0073] As used herein, the term "diagnosis", and variants thereof, such as, but not limited
to "diagnose", "diagnosed" or "diagnosing" shall not be limited to a primary diagnosis
of a clinical state, however should be taken to include any primary diagnosis or prognosis
of a clinical state. For example, the "diagnostic assay" formats described herein
are equally relevant to assessing the remission of a patient, or monitoring disease
recurrence, or tumor recurrence, such as following surgery or chemotherapy, or determining
the appearance of metastases of a primary tumor. All such uses of the assays described
herein are encompassed by the present invention.
[0074] Accordingly, the level of PLA
2 expression, at either the RNA level or the protein level, can be used to diagnose
prostate cancer, or as a prognostic to monitor the progress of prostate cancer.
[0075] Preferred nucleic acid-based diagnostic assays rely upon the detection or relative
quantification of RNA levels in samples using probes of at least about 20 nucleotides
in length that hybridize specifically to RNA, encoding PLA
2, or alternatively, amplify cDNA from RNA encoding PLA
2. Conveniently, any hybridization assay format can be used to detect PLA
2-encoding RNA in samples, such as, for example, high-throughput screening using microarray
technology, or conventional northern hybridization or reverse transcription polymerase
chain reaction (i.e. RT-PCR). In situ localization can also be employed using histology
specimens.
[0076] Suitable diagnostic immunoassays utilize antibodies, including monoclonal and polyclonal
antibodies, or a Fab fragment, F(ab')2 fragment, or scFv fragment, that binds to a
unique peptide region comprising at least about 5-10 contiguous amino acid residues
of PLA
2.
[0077] The present invention further encompasses any synthetic or recombinant peptides,
or antibodies suitable for use in the assays described herein.
[0078] ln preferred embodiments of these diagnostic methods the PLA
2 is sPLA
2-IIA or cPLA
2-É.
[0079] In another aspect, the invention relates to methods of diagnosing for predisposition
to prostate cancer.
[0080] Accordingly, in one aspect the present invention provides a method of assessing the
predisposition of a subject to prostate cancer, the method comprising the step of
determining the presence of a polymorphism or an epigenetic change in a PLA
2 gene of the subject.
[0081] In one embodiment, the polymorphism is a PLA
2 polymorphism already identified in a public or private database such as the NCBI
database or the Celera database.
[0082] The present invention encompasses nucleic-acid based methods and protein-based methods
for diagnosing susceptibility to prostate cancer.
[0083] The polymorphism in the PLA
2 gene may be a point mutation (i.e. a single nucleotide polymorphism (SNP)), deletion
and/or insertion. Such a polymorphism may be detected by isolating and sequencing
DNA fragments from the PLA
2 gene or otherwise by isolating mRNA from the individual and synthesising DNA therefrom
(e.g. by RT-PCR) for sequencing. Polymorphisms may also be detected by hybridisation
using discriminating oligonucleotide probes or by amplification procedures using discriminating
oligonucleotide primers. Suitable methods may involve Southern analysis of genomic
DNA; direct mutation analysis by restriction enzyme digestion; Northern analysis of
RNA; denaturing high pressure liquid chromatography (DHPLC); gene isolation and sequencing;
hybridization of an allele-specific oligonucleotide with amplified gene products;
exon trapping, single base extension (SBE); or analysis of a PLA
2 protein.
[0084] In one embodiment the epigenetic change is aberrant methylation in a PLA
2 gene of the subject or insertion of an endogenous retroviral promoter or transposable
element promoter close to a PLA
2 gene of the subject.
Methods of screening for therapeutic agents
[0085] Also encompassed by the present invention are methods of identifying therapeutic
agents useful for the treatment of prostate cancer.
[0086] Accordingly, in a first aspect the present invention provides a method of screening
for a compound that reduces or inhibits the proliferation of prostate cells, the method
comprising determining the activity of PLA
2 in the presence and absence of a candidate compound, wherein reduced PLA
2 activity in the presence of the compound indicates that the compound reduces or inhibits
the proliferation of prostate cells.
[0087] In another aspect, the present invention provides a method of screening for a compound
that reduces or inhibits the proliferation of prostate cells, the method comprising
determining the expression levels of PLA
2 in the presence and absence of a candidate compound, wherein reduced PLA
2 expression in the presence of the compound indicates that the compound reduces or
inhibits the proliferation of prostate cells.
[0088] In a further embodiment of this aspect, the method involves exposing a translation
system capable of expressing PLA
2 to a candidate compound and comparing the levels of expression of PLA
2 in the presence of the compound to the levels achieved under similar conditions but
in the absence of the compound. The translation system may be a cell-free translation
system. Alternatively, the translation system may comprise eukaryotic or prokaryotic
cells.
[0089] In a further aspect the present invention provides a method of screening for a compound
that reduces or inhibits the proliferation of prostate cells, the method comprising
determining the ability of a candidate compound to modulate the binding of PLA
2 to a PLA
2 substrate, wherein an altered level of binding of PLA
2 to the substrate in the presence of the compound indicates that the compound reduces
or inhibits the proliferation of prostate cells.
[0090] In preferred embodiments of these screening methods the PLA
2 is sPLA
2-IIA or cPLA
2-É.
Therapeutic methods
[0091] We have shown that administration of exogenous sPLA
2-IIA to prostate cells stimulates cell proliferation. We have also shown that administration
of PLA
2 inhibitors inhibits sPLA
2-IIA mediated cell proliferation. Consequently, PLA
2 inhibitors can be used to inhibit or reduce prostate cell proliferation in cells,
particularly in cells with elevated sPLA-IIA activity such as prostate cancer cells.
[0092] The PLA
2 inhibitors can be used therapeutically for prostate cancers, particularly AIPCs.
The term "therapeutically" or as used herein denotes both prophylactic as well as
therapeutic administration. Thus, PLA
2 inhibitors can be administered to high-risk patients in order to lessen the likelihood
and/or severity of prostate cancer or administered to patients already evidencing
prostate cancer.
[0093] The peptides, analogues and small molecule inhibitors described above may preferably
be combined with various components to produce compositions. Preferably the compositions
are combined with a pharmaceutically acceptable carrier or diluent to produce a pharmaceutical
composition (which may be for human or animal use). Suitable carriers and diluents
include isotonic saline solutions, for example phosphate-buffered saline, water, dry
powders and micelles. The composition may be administered by any means known in the
art. Modes of delivery include, but are not limited to, direct injection, topical
delivery (e.g. by atomised nasal delivery or nasal drops) or oral delivery. Accordingly,
the composition may be formulated,
inter alia, for topical, parenteral, intramuscular, intravenous, subcutaneous, intraocular,
oral or transdermal administration.
[0094] Typically, each peptide or analogue or derivative thereof may be administered at
a dose of from 0.01 to 30 mg/kg body weight, preferably from 0.1 to 10 mg/kg, more
preferably from 0.1 to 1 mg/kg body weight.
[0095] The routes of administration and dosages described are intended only as a guide since
a skilled practitioner will be able to determine readily the optimum route of administration
and dosage for any particular patient and condition.
[0096] The present invention will now be described further with reference to the following
examples which are intended to be illustrative only and non-limiting.
EXAMPLES
Materials and Methods
[0097] Cell Lines and Culture. The LNCaP, DU145 and PC3 cell lines were purchased from American Type Culture Collection
(Rockville, MD). All cell cultures were maintained in RPMI 1640 supplemented with
5% FBS for LNCaP and 10% FBS for DU145 and PC3 at 37°C in a humidified environment
of 5% CO
2. For experiments with addition of androgens, cells were cultured in RPMI 1640 without
phenol red and supplemented with same concentration of charcoal stripped FBS. For
experiments described in this particle, we used cells between passage 30 and 45 for
LNCaP, 65 and 80 for DU145 and 25 and 40 for PC3.
[0098] Reagents. sPLA
2-IIA was purified by immunoaffinity chromatography from conditioned media derived
from Chinese hamster ovary cell line (5A2) expressing human sPLA
2-IIA as described (
Bidgood, M.J. et a/. J. Immunol. (2000) 165:2790-2797). A cDNA encoding the activity-dead mutant sPLA
2-IIA H
48Q was constructed by oligonucleotide-mediated site-directed mutagenesis using standard
molecular techniques. This cDNA was expressed in Chinese hamster ovary cells under
the control of the human metallothionein promoter. H
48Q was purified from conditioned media derived from this cell line by immunoaffinity
chromatography. Purity was verified by the presence of a single band on a silver-stained
SDS gel and by amino terminal peptide sequence analysis. Enzyme activity was undetectable
in the purified protein as determined by the radiolabelled
Eschericia coli assay (
Church, W.B. et al. (2001). J. Biol. Chem. 276:33156-33614). Both mutant protein and sPLA
2-IIA were quantified by ELISA (
Smith, G.M. et al., Br. J. Rheumatol. (1992) 31: 175). sPLA
2-IIA inhibitors (Church, W.B. et al) were synthesised using Fmoc (N-(9-fluoroenyl)methoxycarbonyl)
solid phase chemistry without removal of side-chain protection groups prior to cleavage
from the resin. Peptides were cyclised using standard peptide synthesis activation
and coupling chemistry prior to deprotection (Auspep, Melbourne). The cPLA
2 inhibitor pyrrolidone-1 was a kind gift from Dr Michael Gelb.
[0099] Antibodies used in these experiments included polyclonal anti-sPLA
2-IIa (160502, Cayman Chemicals, monoclonal anti-sPLA
2-IIA antibody 4A1 raised by ourselves (Smith, G.M.
et al.), and anti-cPLA
2 polyclonal antibody (SC-438, Santa Cruz).
[0100] Cell growth assay. Cells were plated at 1 x 10
4 per well in 96-well plates with 0.1 ml of FBS-supplemented RPMI. After reaching 70-80%
confluence (about 48 hours), the medium was changed to that containing various treatments
for 3 days. After treatment, the number of viable cells was determined using the MTS
assay (CellTiter 96® AQueous Assay; Promega, Madison, WI). In brief, 20 ul of MTS
solution was added to each well and cells were incubated for 1 h. The absorbance at
490 nm was measured with an ELISA microplate reader. Each experiment was performed
in quadruplicate and repeated at least three times.
[0101] Flow Cytometric Analysis. Cells were seeded in 25 ml flasks in conditions as described above. Following treatment,
trypsinization and cell counting, LNCaP cells (1x10
6) were suspended in 1 mL PBS and incubated with 0.2 mL 0.4 % Triton X-100 for 5 min
at R/T in the presence of 50 µL of propidium iodide solution (50 µg/mL) and 20 µL
of ribonuclease (10 mg/mL). DNA content per cell was measured by flow cytometry using
an FACScalibur flow cytometer and CellQuest software (Becton Dickinson, Franklin Lakes,
NJ). Statistical analysis was performed on 10,000 events per sample.
[0102] RT-PCR. Total cellular RNA was isolated from LNCaP, DU145 or PC-3 cells using the Trizol
reagent (LifeTechnologies, Inc.). First-strand cDNA was synthesized from 5 µg of RNA
with the cDNA preamplification system (Life Technologies, Inc.) using SuperScript
II reverse transcriptase and an oligo(dT) primer. This was used as the template in
standard PCR reactions using Amplitaq DNA polymerase (Perkin-Elmer Life Sciences,
Boston, MA). Amplification products were analyzed on 2% TAE agarose gels made with
MetaPhor agarose (FMC BioProducts, Rockland, ME) and photographed under UV illumination.
DNA ladders of 25 and 100 bp (Life Technologies, Inc.) were used as size standards.
Primers were designed based on the human sPLA
2-IIA mRNA (NM_000300.2) deposited in the GenBank database (National Center for Biotechnology
Information, Bethesda, MD) and are as follows: forward: 5'-TTTGTCACCCAAGAACTCTTAC-3',
reverse: 5'-GGGAGGGAGGGTATGAGA-3'.
[0103] Tissues. Normal prostate was obtained from brain-dead organ donors as described previously
(
Chetcuti et al., (2001) Cancer Research, 61:6331-6334). Benign prostatic hyperplasia (BPH) was obtained from transurethral resection of
the prostate. Cancer tissue was from radical prostatectomy. Informed consent was obtained
from patients, and the study protocol was approved by the Central Sydney Area Health
Service Ethics Review Committee.
[0104] Immunohistochemistry. Prostate tissues (normal, benign hyperplastic and cancer) were all fixed in 10% formaldehyde
solution for <24 hours and paraffin embedded. Tissue sections (5 µm) were incubated
for 1 hour at 37°C after microwave antigen retrieval with the appropriate primary
antibody diluted in 1% goat serum. Biotinylated goat anti-rabbit or mouse IgG, diluted
1/200 in 1% normal goat serum, was used as the secondary antibody. The signal was
amplified using the avidin-biotin-peroxidase complex system (Vector Laboratories)
and visualised using the liquid DAB substrate-chromogen system (Dako). Sections were
counterstained with hematoxylin. Negative isotype and method controls were performed
for each sample by substituting the primary antibody with rabbit IgG and 1% goat serum,
respectively.
[0105] Image Analysis. Immunoperoxidase staining was considered positive and specific when the intensity
of with the post-immune IgG clearly exceeded that observed with the preimmune IgG
(isotype control) and with no IgG (method control). Specific staining was graded for
the percentage of immunopositive normal or cancer epithelial cells as described (
Kommoss, 1989 Anal. Quant. Cytol. Histol. 11:298-306). Briefly, the percentage of positive-stained normal, or cancerous epithelial cells
per slide was stratified into three groups: group 1, 0-33%; group 2, 34-66%; and group
3, 67-100%. Zero represented no cells with specific staining.
[0106] Statistical Analysis. The Number Cruncher Statistical System (NCSS, Kaysville, UT) was used for statistical
analysis. Data were analyzed by one-way ANOVA and correlation coefficient as appropriate.
A two-tailed P value <0.05 was considered significant.
Example 1: sPLA2-IIA is Induced in Prostate Cancer Cells and Constitutively Activated in AIPC.
[0107] We searched the database of Serial Analysis of Gene Expression (SAGE) to determine
the steady state mRNA levels of individual PLA
2 enzymes in prostate cancer. The cDNA libraries used for SAGE was PR317 normal prostate
and PR317 prostate cancer, respectively (www.ncbi.nlm.nih.gov/SAGE), as both are derived
from microdissected prostate tissues. We found that sPLA
2-IIA mRNA. was 22 times higher in prostate cancer than normal prostate, whereas other
members were either not expressed in the prostate libraries or unchanged in cancer.
To verify the SAGE result and extend the expression analysis to androgen-independent
prostate cancer (AIPC), we examined sPLA
2-IIA expression by immunohistochemistry in prostate cancer tissues from patients treated
with androgen-ablation therapy for 3 months prior to radical prostatectomy. Cancer
cells remaining in specimens following androgen-ablation therapy are regarded as being
closest to AIPC, although they are confined within the prostate. Cancer specimens
from patients undergoing radical prostatectomy without androgen ablation therapy served
as the control. Two antibodies were used for immunohistochemistry, and both showed
the same expression pattern. In the control group, (N=50), there was weak and patchy
staining in benign glands (Fig. 1A and C.) adjacent to cancer cells and extensive
staining in cancer cells (Fig. 1C.). In the androgen-ablated group (N=25), benign
glands lost their staining, whereas AIPC cells maintained sPLA
2-IIA expression (Fig. 1B and C). We also found that the extent of sPLA
2-IIA staining is positively correlated with the tumour grade and post-operative PSA
level (data not shown). The chromosomal location of sPLA
2-IIA (1p35.1-36) was also found to overlap with a prostate cancer susceptibility locus
CAPB (
Gibbs et al (1999) Am. J. Hum. Genet. 64:776-787). No difference was found in immunohistochemical staining for cPLA
2-α between normal and cancer cells irrespective of androgen status (data not shown).
[0108] The lack of sPLA
2-IIA expression in benign glands following androgen deprivation suggests that expression
of sPLA
2-IIA gene requires androgens. To verify that, we searched the 5'- flanking region
of the sPLA
2-IIA gene using MatInspector Release 5.3 (Genomatix), and found an androgen response
element (ARE) GAGGTAAATGGTATTCTC from -546 to -527. Secondly, we treated the androgen
responsive human prostate cancer cell line, LNCaP, with various doses of androgens
and measured sPLA
2-IIA mRNA and protein levels by RT-PCR and ELISA (data not shown). Indeed, there was
an increase in the level of sPLA
2-IIA transcript following 1 nM androgen treatment. In contrast, we could not find
an ARE within 3 kb of genomic DNA in the 5-flanking region of the cPLA
2-α gene. Androgen treatment had no effect on cPLA
2-α mRNA and protein levels (data not shown). Together, these findings indicate that
sPLA
2-IIA, but not cPLA
2-α expression is normally dependent on androgens and in AIPC sPLA
2-IIA expression becomes androgen-independent
via an as yet unknown mechanism.
Example 2: Oncogenic Action of sPLA2-IIAin Prostate Cancer Cells.
[0109] To examine the biological relevance of sPLA
2-IIA to prostate cancer cell growth, we treated LNCaP cells with increasing doses
of human recombinant sPLA
2-IIA and monitored cell growth. LNCaP is the only available prostate cancer cell line
that expresses both the androgen receptor and PSA. Exogenously-added sPLA
2-IIA at doses as low as 1 nM, consistently and dose-dependently stimulated LNCaP cell
growth as measured by the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethylphenyl)-2-(4-sulphophenyl)-2H-tetrazolium
(MTS) assay (Fig. 2A). In contrast addition of an activity-dead sPLA
2-IIA mutant protein H
48Q, showed no stimulation relative to untreated cells (Fig. 2A) indicating that enzyme
activity is essential for the growth-stimulatory effect.
[0110] We have previously shown that human sPLA
2-IIA is dose-dependently inhibited by a pentapeptide sequence comprising residues
70-74 of the native sPLA
2-IIA protein (
70FLSYK
74) (
Tseng. A., et al., (1996) J. Biol. Chem. 271:23992-23998). Because of the inherent flexibility of the linear peptide sequence, inhibition
was weak in
in vitro activity assays. We have recently designed two novel cyclic peptides (Church, W.B.
et al.), cFLSYR and a cyclic peptide where F and Y are substituted with 2-naphthylalanine
(c(2NapA)LS(2NapA)R). Both have shown significant improvement in potency over linear
peptides. The potent stimulatory effect of exogenous sPLA
2-IIA on prostate cancer cell number was completely blocked by the sPLA
2-IIA inhibitor, cFLSYR (Fig. 2B) at all concentrations tested.
[0111] We next used flow cytometric analysis to determine how sPLA
2-IIA affects the distribution of LNCaP cells in different phases of the cell cycle.
In sPLA
2-IIA containing medium, the proportion of LNCaP cells in the G1 phase decreased from
74% to 62% with corresponding increase of cells in G2/M phase in comparison to untreated
cells (Fig. 2C). In the presence of both sPLA
2-IIA and its inhibitor (cFLSYR), the proportion of cells in G1 and G2 phase returned
to basal levels (Fig. 2C). These results establish the biological importance of the
sPLA
2-IIA enzyme activity in prostate cancer cells and demonstrate that sPLA
2-IIA-induced cell growth can be attributed at least partly to an increased proportion
of cells entering G2/M phase from G1 phase.
Example 3: Oncogenic Action of sPLA2-IIA requires cPLA2-α activity.
[0112] Currently, two models are proposed to explain the action of sPLA
2 on eicosanoid production (
Murakami, M., Kudo, I. (2002) J. Biochem. 131:285-292). One is the direct cleavage of membrane phospholipids. In this model, sPLA
2 binds directly to plasma membrane phospholipids and the released arachidonic acid
serves as a substrate to produce eicosanoids. Lysophospholipids and/or eicosanoid
products of arachidonic acid metabolism mediate indirect activation of the endogenous
cPLA
2-α via mobilisation of calcium. The enhanced cPLA
2-α activity can in turn result in enhanced production of eicosanoids. The second model
is indirect modulation of intracellular eicosanoid pathways
via cell surface GPI-linked heparan sulphate proteoglycan receptors. The internalised
sPLA
2 supplies arachidonic acid to downstream enzymes either directly, or indirectly via
activation of cPLA
2-α through mitogen activated protein (MAP) kinase-mediated phosphorylation.
[0113] To determine if sPLA
2-IIA-induced cell proliferation depends on cPLA
2-α, we treated LNCaP cells with various doses of selective cPLA
2 inhibitor, pyrrolidine-1 (
Ghomashahi, F. et al. (2001) Biochim. Biophys. Acta Biomembranes 2:160-166) with or without a constant effective dose of sPLA
2-IIA (1 nM). Blockade of cPLA
2-α abolishes sPLA
2-IIA-induced cell growth completely (Fig. 3), demonstrating that cPLA
2-α activation is necessary for sPLA
2-IIA-dependent cell proliferation. In the absence of sPLA
2-IIA, the cPLA
2-α inhibitor had no effect on cell growth, suggesting that cPLA
2-α does not promote cell proliferation independently in LNCaP cells (see legend Fig.
3).
Example 4: sPLA2-IIA Inhibitors Suppress Endogenous Proliferation.
[0114] Based on our finding that the growth-promoting sPLA
2-IIA is constitutively expressed in AIPC we have considered the potential of sPLA
2-IIA as a target for treatment of AIPC. We reason that a better outcome can be achieved
with the PLA
2 inhibitor than with a COX inhibitor alone because the latter suppresses the production
of prostaglandins only.
[0115] To test the effect of blocking endogenous sPLA
2-IIA on cell growth, we firstly determined the basal mRNA levels of sPLA
2-IIA in 3 human prostate cancer cell lines. The androgen-independent cell lines PC-3
and DU145 cells were included in the study to also exclude a possibly general toxicity
of the inhibitors. mRNA encoding sPLA
2-IIA was undetectable in DU-145 compared with LNCaP and PC-3 cells (Fig 4A). We then
tested the effect of individual inhibitors cFLSYR and c(2Nap)LS(2Nap)R on cell growth
over a range of doses (1-100 nM). The proliferation of LNCaP and PC-3 cells was significantly
decreased, and the smallest effective dose was 1 nM for both inhibitors (Fig. 4B).
In contrast, neither of the inhibitors had an effect on DU145, presumably due to the
lack of endogenous sPLA
2-IIA. The low but effective dosage and its specificity for cell lines containing endogenous
sPLA
2-IIA only, indicate that non-specific cell toxicity is not likely to explain the inhibitory
effect.
[0116] In summary, the normally androgen-induced sPLA
2-IIA gene expression is constitutively activated in androgen-independent prostate
cancer. Exogenously added sPLA
2-IIA promotes prostate cell proliferation through its enzyme product and cPLA
2. A better therapeutic outcome is likely to be achieved with an sPLA
2 inhibitor in prostate cancer including the form of AIPC.